Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Aesthetic Plast Surg ; 47(1): 465-472, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36536095

RESUMEN

BACKGROUND: There is no consensus regarding the choice of injected drugs for pathological scars. Although the clinical efficacy of different drug treatments was shown in many randomized controlled trials, the efficacies of many drugs are inconsistent. Therefore, this study aimed to determine how different effective drugs are for treating pathological scars. It is anticipated that the study findings may serve as guidelines for plastic surgeons. METHODS: Relevant literature was extracted from the following databases Cochrane Library, Embase, PubMed, Web of Science, CNKI, Weipu, and Wanfang until June 2022, such as randomized clinical trials (RCTs) evaluating different injected drugs for the treatment of pathological scars, including BTA, TAC, 5-Fu, VER, and BLE. RESULTS: This network meta-analysis of 1539 patients from 23 articles revealed that the most effective treatment for a pathological scar was TAC + BTA. The effective rate of TAC + BTA combination therapy was significantly different from that of the BTA, TAC, 5-Fu, VER, and BLM monotherapies. TAC+5-FU was more effective than TAC, 5-FU, VER, or BLM alone, and BTA was more effective than both TAC and 5-Fu. The effectiveness of VER and BLM was the same, but both were better than TAC and 5-Fu. No big differences were found between any of the other local injection therapies. CONCLUSIONS: According to this network meta-analysis, a combination of keloid and hypertrophic scar injection treatment is recommended, especially BTA+TAC. However, this network meta-analysis has some limitations and must be further verified by larger samples and higher quality RCTs. LEVEL OF EVIDENCE III: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.


Asunto(s)
Cicatriz Hipertrófica , Queloide , Humanos , Queloide/patología , Cicatriz Hipertrófica/patología , Metaanálisis en Red , Resultado del Tratamiento , Fluorouracilo/uso terapéutico
2.
Biomater Res ; 26(1): 83, 2022 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-36528594

RESUMEN

BACKGROUND: Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been found to have a great potential for soft tissue repair due to various biological functions, including pro-angiogenesis and low immunogenicity. However, the low yield and heterogeneity of MSC-EVs limited their clinical transformation. This study was designed to develop a novel adipose-derived stem cell engineered nanovesicles (ADSC-NVs) with high production and explore its pro-angiogenetic effect and application in adipose tissue regeneration. METHODS: Adipose-derived stem cell-derived extracellular vesicles (ADSC-EVs) were isolated from an EVs-free culture medium for human ADSCs (hADSCs). ADSC-NVs were prepared by sequentially extruding ADSCs followed by iodixanol density gradient ultracentrifugation and were compared with ADSC-EVs in morphology, size distribution, protein contents and yield. The pro-angiogenetic effect of ADSC-NVs in different doses (0, 5, 20 and 80 µg/mL) in vitro was determined using transwell assay, tube formation assay, western blot and qRT-PCR. In vivo, BALB/c nude mice were administered injection of a mixture of fat granules and different dose of ADSC-NVs and grafts were harvested at 12 weeks post-transplantation for further analysis. By analyzing the weight and volume of grafts and histological evaluation, we investigated the effect of ADSC-NVs in vessel formation and adipose tissue regeneration. RESULTS: Our results showed yield of purified ADSC-NVs was approximately 20 times more than that of ADSC-EVs secreted by the same number of ADSCs. In vitro, both ADSC-NVs and ADSC-EVs exhibited a dose-dependent pro-angiogenetic effect, despite their distinct miRNA profiles. These effects of ADSC-NVs may be mediated by enriched miR-21-5p via PTEN inhibition and PI3K/p-Akt signaling activation. Furthermore, after a mixed injection of ADSC-NVs, vessel formation and adipose regeneration were observed in vivo in fat implants. CONCLUSIONS: Our study developed a potent alternative of ADSC-EVs. ADSC-NVs have a high pro-angiogenesis potential and can be used as cell-free therapeutic biomaterials in soft tissue regeneration.

3.
Regen Biomater ; 8(2): rbab003, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33738117

RESUMEN

Associated with persistent oxidative stress, altered inflammatory responses, poor angiogenesis and epithelization, wound healing in diabetic patients is impaired. N-acetylcysteine (NAC) is reported to resist excess reactive oxygen species (ROS) production, prompt angiogenesis and maturation of the epidermis. Studies have revealed that graphene oxide (GO) can regulate cellular behavior and form cross-links with naturally biodegradable polymers such as collagen (COL) to construct composite scaffolds. Here, we reported a COL-based implantable scaffold containing a mixture of GO capable of the sustained delivery of NAC to evaluate the wound healing in diabetic rats. The morphological, physical characteristics, biocompatibility and NAC release profile of the GO-COL-NAC (GCN) scaffold were evaluated in vitro. Wound healing studies were performed on a 20 mm dorsal full-skin defect of streptozotocin (STZ)-induced diabetic rats. The injured skin tissue was removed at the 18th day post-surgery for histological analysis and determination of glutathione peroxidase (GPx), catalase (CAT) and superoxide dismutase (SOD) activity. In diabetic rats, we confirmed that the GCN scaffold presented a beneficial effect in enhancing the wound healing process. Additionally, due to the sustained release of NAC, the scaffold may potentially induce the antioxidant defense system, upregulating the expression levels of the antioxidant enzymes in the wound tissue. The findings revealed that the antioxidant biocompatible composite collagen dressing could not only deliver NAC in situ for ROS inhibition but also promote the wound healing process. This scaffold with valuable therapy potential might enrich the approaches for surgeon in diabetic wound treatment in the future.

4.
J Plast Reconstr Aesthet Surg ; 73(7): 1318-1325, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32430265

RESUMEN

BACKGROUND: Application of distant skin flaps in facial defect reconstruction has limitations such as leaving a patch like appearance and being restricted by the length of the vascular pedicles. Leveraging the abundance of blood supply from superficial muscular aponeurotic system (SMAS), a local skin flap pedicled by SMAS can be used to avoid the aforementioned problems. Herein, we report the clinical application as well as the anatomical study of SMAS-pedicled skin flaps. METHODS: This study enrolled patients who underwent facial defect reconstruction surgery between 2013 and 2018 using SMAS-pedicled skin flaps. The flaps were designed according to the size and location of the defect. A follow-up was performed to evaluate the treatment outcomes and incidence of adverse events. In addition, six cadaveric heads were used to perform an anatomical study on the distribution and blood supply of SMAS. RESULTS: Twenty-three cases underwent the defect reconstruction surgery in the frontal regions (three cases), temporal region (four cases), periocular region (four cases), nasal region (seven cases), and other regions (five cases). All the flaps survived well. During the follow-up period up to 12 months, the flaps showed a satisfactory appearance, blood supply, and elasticity. The distribution and blood supply of SMAS at different anatomical regions have been successfully observed. Abundant vascular networks could be found in the SMAS layer. CONCLUSION: Based on the broad distribution of SMAS and the abundant blood supply, an SMAS-pedicled skin flap could be flexibly designed and versatilely used to reconstruct post-traumatic or post-excisional facial defects.


Asunto(s)
Aponeurosis/anatomía & histología , Cara/cirugía , Procedimientos de Cirugía Plástica/métodos , Colgajos Quirúrgicos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Aponeurosis/irrigación sanguínea , Aponeurosis/trasplante , Cadáver , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
5.
J Biomed Mater Res A ; 108(12): 2460-2472, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32419333

RESUMEN

Researches of biomaterials for osteoporotic bone defects focus on the improvement of its anti-osteoporosis ability, due to osteoporosis is a kind of systemic and long-range bone metabolism disorder. Nevertheless, how to steadily deliver anti-osteoporosis drugs in osteoporotic bone defects is rarely studied. Reported evidences have shown that alendronate (Aln) is known to not only restrain osteoclasts from mediating bone resorption but also stimulate osteoblasts to regenerate bone tissue. Here, we developed an engineered implantable scaffold that could sustainably release Aln for osteoporotic bone defects. Briefly, Aln was added into 2% collagen (Col) solution to form a 5 mg/ml mixture. Then the mixture was filled into pre-designed round models (diameter: 5 mm, height: 2 mm) and crosslinked to obtain engineered Col-Aln scaffolds. The release kinetics showed that Aln was released at an average rate of 2.99 µg/d in the initial 8 days and could sustainably release for 1 month. To detect the repair effects of the Col-Aln scaffolds for osteoporotic defects, the Col and Col-Aln scaffolds were implanted into 5 mm cranial defects in ovariectomized rats. After 3 months, the cranial defects implanted with Col-Aln scaffolds achieved more bone regeneration in defect area (11.74 ± 3.82%) than Col scaffold (5.12 ± 1.15%) (p < .05). Moreover, ovariectomized rats in Col-Aln scaffold group possessed more trabecular bone in femur metaphysis than Col scaffold group as analyzed by Micro-CT. This study demonstrated the engineered Col-Aln scaffold has the potential to repair osteoporotic bone defects and resist bone loss in osteoporosis.


Asunto(s)
Alendronato , Colágeno , Osteoblastos , Osteoporosis , Alendronato/química , Alendronato/farmacocinética , Alendronato/farmacología , Animales , Colágeno/química , Colágeno/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Femenino , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología , Ovariectomía , Ratas , Ratas Sprague-Dawley
6.
Wound Repair Regen ; 28(1): 126-144, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509318

RESUMEN

Human skin wound repair may result in various outcomes with most of them leading to scar formation. Commonly seen in many cutaneous wound healing cases, hypertrophic scars are considered as phenotypes of abnormal wound repair. To prevent the formation of hypertrophic scars, efforts have been made to understand the mechanism of scarring following wound closure. Numerous in vivo and in vitro models have been created to facilitate investigations into cutaneous scarring and the development of antiscarring treatments. To select the best model for a specific study, background knowledge of the current models of hypertrophic scars is necessary. In this review, we describe in vivo and in vitro models for studying hypertrophic scars, as well as the distinct characteristics of these models. The choice of models for a specific study should be based on the characteristics of the model and the goal of the study. In general, in vivo animal models are often used in phenotypical scar formation analysis, development of antiscarring treatment, and functional analyses of individual genes. In contrast, in vitro models are chosen to pathway identification during scar formation as well as in high-throughput analysis in drug development. Besides helping investigators choose the best scarring model for their research, the goal of this review is to provide knowledge for improving the existing models and development of new models. These will contribute to the progress of scarring studies.


Asunto(s)
Cicatriz Hipertrófica/patología , Modelos Animales de Enfermedad , Técnicas In Vitro , Piel/patología , Animales , Técnicas de Cultivo de Célula , Cicatriz Hipertrófica/metabolismo , Cricetinae , Cobayas , Humanos , Ratones , Modelos Teóricos , Conejos , Ratas , Piel/metabolismo , Porcinos
7.
Biofabrication ; 12(1): 015023, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31665714

RESUMEN

Breast tissue engineering is a promising alternative to standard treatments for breast defects. Although there is a consensus that the mechanical property of the scaffold should best match the reconstructed tissue, the simulation of the soft and elastic tactility of native breast tissues using conventional materials and architecture design requires further study. Previous research has shown that the crystal microstructure-like design can drastically alter the mechanical properties of the constructed scaffolds. In this study, we designed and additive manufactured four kinds of breast scaffolds using polyurethane and termed their architectures as N5S4, N9S8, N7S6 and N4S6. The basic unit cell of each scaffold was similar to a lattice structure from the isometric crystal system. The scaffolds possessed identical porosity but different mechanical properties in which the compressive modulus of the softest scaffolds (N5S4) were similar to that of native breast tissue. When applied in the construction of tissue-engineered breast combining with delayed fat injection technique in nude rat models, the soft scaffolds(N5S4) performed better compared to its stiff counterpart (N4S6), as higher adipose survival, vascularization and milder fibrosis could be observed in N5S4 scaffolds . Lastly, using finite element analysis, we further investigated the influence of the unit cell architectures on the mechanical properties of the scaffolds and simulated the deformation as well as stress distribution patterns of the implanted scaffolds in detail. Thus, a crystal lattice-like architecture design was introduced to tune the mechanical properties of the scaffolds and match the requirements for tissue engineering applications.


Asunto(s)
Mama/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Bioimpresión , Mama/citología , Femenino , Porosidad , Impresión Tridimensional , Ratas , Ratas Desnudas , Ingeniería de Tejidos/instrumentación
8.
Theranostics ; 9(20): 5839-5853, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534523

RESUMEN

Wound dressings composed of natural polymers, such as type I collagen, possess good biocompatibility, water holding capacity, air permeability, and degradability, and can be used in wound repair. However, due to the persistent oxidative stress in the wound area, the migration and proliferation of fibroblasts might be suppressed, leading to poor healing. Thus, collagen-containing scaffolds are not suitable for accelerated wound healing. Antioxidant N-acetyl cysteine (NAC) is known to reduce the reactive oxygen species (ROS) and has been widely used in the clinic. Theoretically, the carboxyl group of NAC allows loading of graphene oxide (GO) for sustained release and may also enhance the mechanical properties of the collagen scaffold, making it a better wound-dressing material. Herein, we demonstrated an innovative approach for a potential skin-regenerating hybrid membrane using GO incorporated with collagen I and NAC (N-Col-GO) capable of continuously releasing antioxidant NAC. Methods: The mechanical stability, water holding capacity, and biocompatibility of the N-Col-GO hybrid membrane were measured in vitro. A 20 mm rat full-skin defect model was created to evaluate the repair efficiency of the N-Col-GO hybrid membrane. The vascularization and scar-related genes in the wound area were also examined. Results: Compared to the Col only scaffold, N-Col-GO hybrid membrane exhibited a better mechanical property, stronger water retention capacity, and slower NAC release ability, which likely promote fibroblast migration and proliferation. Treatment with the N-Col-GO hybrid membrane in the rat wound model showed complete healing 14 days after application which was 22% faster than the control group. HE and Masson staining confirmed faster collagen deposition and better epithelization, while CD31 staining revealed a noticeable increase of vascularization. Furthermore, Rt-PCR demonstrated decreased mRNA expression of profibrotic and overexpression of anti-fibrotic factors indicative of the anti-scar effect. Conclusion: These findings suggest that N-Col-GO drug release hybrid membrane serves as a better platform for scarless skin regeneration.


Asunto(s)
Acetilcisteína/química , Colágeno/química , Grafito/química , Acetilcisteína/uso terapéutico , Animales , Movimiento Celular/efectos de los fármacos , Módulo de Elasticidad , Masculino , Ratones , Microscopía Electrónica de Rastreo , Células 3T3 NIH , Porosidad , Ratas , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Difracción de Rayos X
9.
J Biomed Mater Res A ; 107(7): 1414-1424, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30737888

RESUMEN

PCL (poly-caprolactone) nanofibers have good biocompatibility and high porosity, which are usually utilized for application in wound dressings. However, wound healing could be hindered by the overproduction of reactive oxygen species (ROS) and different factors. Pure nanofibers cannot satisfy these requirements of wound healing. N-acetylcysteine (NAC), as an antioxidant, meets the requirements for wound healing by resisting the overproduction of ROS and by promoting angiogenesis and maturation of the epidermis. In this study, we prepared a sandwich structured PCL-Col/NAC scaffold using the molding method, which consisted of PCL nanofibers at the core and NAC-loaded collagen on both sides. The hydroscopicity and tensile modulus of PCL-Col/NAC scaffolds showed best performance of these properties among groups. Meanwhile, the drug release profiles of PCL-Col/NAC scaffolds were investigated using the HPLC method and the results suggested a sustained drug release of NAC for PCL-Col/NAC scaffolds. In addition, PCL-Col/NAC scaffolds presented better properties than the control groups in cell migration and proliferation. The in vivo wound healing therapy effect was studied using an oval (2 × 1 cm) full-thickness skin defect wound model for SD rats. After 21 days, gross view and histological analysis showed a favorable beneficial therapeutic effect as well as better epidermal maturation compared with the control groups. CD31 immunohistology results revealed relatively more new vessels in the PCL-Col/NAC group than the control groups. This study developed novel PCL-Col/NAC scaffolds with an excellent hydroscopicity, tensile modulus and the ability to promote epidermal maturation and angiogenesis, demonstrating its promising potential in wound healing treatment. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A, 2019.


Asunto(s)
Acetilcisteína/farmacología , Colágeno/química , Poliésteres/química , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Materiales Biocompatibles/farmacología , Movimiento Celular/efectos de los fármacos , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Ratones , Células 3T3 NIH , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Implantación de Prótesis , Ratas Sprague-Dawley , Resistencia a la Tracción , Factores de Tiempo
10.
J Biomed Mater Res A ; 107(3): 678-688, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30474182

RESUMEN

Tissue-engineered bone grafts (TEBGs) represent a promising treatment for bone defects. Nevertheless, drawbacks of the current construction strategy (top-down [TD] strategy) such as limited transmission of nutrients and nonuniform distribution of seeded cells, result in an unsatisfied therapeutic effect on large segmental bone defects. Theoretically, tissue-engineered microtissue (TEMT)-based bottom-up (BU) strategy is effective in preserving seed cells and vascularization, thus being regarded as a better alternative for TEBGs. Yet, there are few studies focusing on the comparison of the in vivo performance of TEBGs fabricated by TD or BU strategy. Here, we developed an ectopic bone formation rat model to compare the performance of these two construction strategies in vivo. TEBGs made from gelatin TEMT (BU strategy) and bulk tissue (BT; TD strategy) were seeded with equal number of rat bone marrow-derived mesenchymal stem cells and fabricated in 5 mm polydimethylsiloxane chambers. The grafts were implanted into subcutaneous pockets in the same rat. Four weeks after implantation, microcomputed tomography and hematoxylin and eosin staining results demonstrated that more bony tissue was formed in the microtissue (MT) group than in the BT group. CD31 staining further confirmed that there were more blood vessels in the MT group, indicating that the BU strategy was superior in inducing angiogenesis. This comparative study provides evidence that the BU construction strategy is more effective for in vivo application and bone defect treatment by bone tissue engineering. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 678-688, 2019.


Asunto(s)
Bioprótesis , Células de la Médula Ósea/metabolismo , Huesos , Gelatina/química , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Tejidos , Animales , Células de la Médula Ósea/citología , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley
11.
ACS Appl Mater Interfaces ; 10(50): 44080-44091, 2018 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-30475576

RESUMEN

Biomimetic mineralization using simulated body fluid (SBF) can form a bonelike apatite (Ap) on the natural polymers and enhance osteoconductivity and biocompatibility, and reduce immunological rejection. Nevertheless, the coating efficiency of the bonelike apatite layer on natural polymers still needs to be improved. Graphene oxide (GO) is rich in functional groups, such as carbonyls (-COOH) and hydroxyls (-OH), which can provide more active sites for biomimetic mineralization and improve the proliferation of the rat bone marrow stromal cells (r-BMSCs). In this study, we introduced 0%, 0.05%, 0.1%, and 0.2% w/v concentrations of GO into collagen (Col) scaffolds and immersed the fabricated scaffolds into SBF for 1, 7, and 14 days. In vitro environment scanning electron microscopy (ESEM), energy-dispersive spectrometry (EDS), thermogravimetric analysis (TGA), micro-CT, calcium quantitative analysis, and cellular analysis were used to evaluate the formation of bonelike apatite on the scaffolds. In vivo implantation of the scaffolds into the rat cranial defect was used to analyze the bone regeneration ability. The resulting GO-Col-Ap scaffolds exhibited a porous and interconnected structure coated with a homogeneous distribution of bonelike apatite on their surfaces. The Ca/P ratio of 0.1% GO-Col-Ap group was equal to that of natural bone tissue on the basis of EDS analysis. More apatites were observed in the 0.1% GO-Col-Ap group through TGA analysis, micro-CT evaluation, and calcium quantitative analysis. Furthermore, the 0.1% GO-Col-Ap group showed significantly higher r-BMSCs adhesion and proliferation in vitro and more than 2-fold higher bone formation than the Col-Ap group in vivo. Our study provides a new approach of introducing graphene oxide into bone tissue engineering scaffolds to enhance biomimetic mineralization.


Asunto(s)
Materiales Biomiméticos , Regeneración Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Colágeno , Grafito , Cráneo , Andamios del Tejido/química , Animales , Apatitas/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Colágeno/química , Colágeno/farmacología , Grafito/química , Grafito/farmacología , Ratas , Ratas Sprague-Dawley , Cráneo/diagnóstico por imagen , Cráneo/lesiones , Cráneo/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Microtomografía por Rayos X
12.
ACS Appl Mater Interfaces ; 10(49): 42948-42958, 2018 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-30421913

RESUMEN

Hydrogels such as type I collagen (COL) have been widely studied in bone tissue repair, whereas their weak mechanical strength has limited their clinical application. By adding graphene oxide (GO) nanosheets, researchers have successfully improved the mechanical properties and biocompatibility of the hydrogels. However, for large bone defects, the osteoinductive and cell adhesion ability of the GO hybrid hydrogels need to be improved. Mesenchymal stem cell (MSC) secreted extracellular matrix (ECM), which is an intricate network, could provide a biomimetic microenvironment and functional molecules that enhance the cell proliferation and survival rate. To synergize the advantages of MSC-ECM with GO-COL hybrid implants, we developed a novel ECM scaffold construction method. First, an osteoinductive extracellular matrix (OiECM) was created by culturing osteodifferentiated bone marrow mesenchymal stem cells (BMSCs) for 21 days. Then, the GO-COL scaffold was fully wrapped with the OiECM to construct the OiECM-GO-COL composite for implantation. The morphology, physical properties, biocompatibility, and osteogenic performance of the OiECM-GO-COL implants were assessed in vitro and in vivo (5 mm rat cranial defect model). Both gene expression and cell level assessments suggested that the BMSCs cultured on OiECM-GO-COL implants had a higher proliferation rate and osteogenic ability compared to the COL or GO-COL groups. In vivo results showed that the OiECM-GO-COL implants achieved better repair effects in a rat critical cranial defect model, whereas bone formation in other groups was limited. This study provides a promising strategy, which greatly improves the osteogenic ability and biocompatibility of the GO hydrogels without the procedure of seeding and culturing MSCs on scaffolds in vitro, demonstrating its potential as an off-the-shelf method for bone tissue engineering.


Asunto(s)
Materiales Biomiméticos , Regeneración Ósea/efectos de los fármacos , Matriz Extracelular/química , Grafito , Osteogénesis/efectos de los fármacos , Cráneo , Andamios del Tejido/química , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Femenino , Grafito/química , Grafito/farmacología , Ratas , Ratas Sprague-Dawley , Cráneo/lesiones , Cráneo/metabolismo , Cráneo/fisiología
13.
Ann Plast Surg ; 81(6S Suppl 1): S59-S65, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30418281

RESUMEN

BACKGROUND: Facial aging is a complex process influencing every layer of the facial structure. Most accepted surgical techniques for facial rejuvenation involve certain manipulation of the superficial musculoaponeurotic system (SMAS). Out of these SMAS-based techniques, SMAS plication or suspension provides excellent outcomes with shorter convalescence and fewer potential complications. Herein, we would like to present our own technique combining SMAS plication, periauricular purse-string, and malar fat pad elevation technique for mid and lower facelift. METHODS: Through a classical periauricular and temporal incision, a periauricular permanent purse-string suture was woven into the SMAS to suspend sagging soft tissue of the mid and lower face after superficial undermining, then plication of inner and outer SMAS of the purse-string loop was performed to further secure suspension, and at last the malar fat pad was elevated for midface rejuvenation. The shape of the loop varies with patients' age; for younger patients, the loop is more vertical, and for older patients, the loop is more horizontal. Patient-reported outcomes were described using the FACE-Q questionnaire. RESULTS: From January 2010 to June 2015, a total of 138 patients were treated with this technique by a same surgeon. Follow-up duration ranged from 1 to 6 years. Preoperative and postoperative photographs were recorded and analyzed. The complications rates were low, and satisfaction rates were high. Patients felt that they appeared 7.3 years younger than their actual age on average and were most satisfied with the appearance of their lower face and jawline. CONCLUSIONS: Periauricular purse-string reinforced with SMAS plication and malar fat pad elevation technique produces esthetically pleasing outcomes, besides being simple, safe, and personalized.


Asunto(s)
Tejido Adiposo/cirugía , Cara/cirugía , Ritidoplastia/métodos , Sistema Músculo-Aponeurótico Superficial/cirugía , Técnicas de Sutura , Adulto , Anciano , Mejilla/cirugía , Femenino , Humanos , Persona de Mediana Edad , Rejuvenecimiento
14.
Wound Repair Regen ; 25(5): 858-863, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-29052949

RESUMEN

Diabetic foot ulcer (DFU) is a common complication observed in diabetic patients and affects diabetic patients in multiple ways. Severe DFU even leads to amputation in many cases. Early detection and intervention of DFU in diabetic patients can significantly relieve the pain caused by the ulcer and also keep patients from losing limbs in severe cases. In this study, the risks of diabetic patients getting DFU were estimated through a hospital-based survey. This survey collected information from hospitalized diabetic patients in Wuhan City, Hubei Province, China, using a questionnaire. This investigation includes studies from two stages with 502 diabetic patients from 20 hospitals in Wuhan City. The results suggested that patients with a long history of diabetes are often associated with a high risk of DFU (χ2 = 11.428, p = 0.0007), smoking (χ2 = 8.386, p = 0.0007), diabetic complications (χ2 = 13.484, p < 0.0001), and especially patients with diabetic foot complications (χ2 = 57.6621, p < 0.0001). Foot lesions appeared to be important attributors to DFU since our data demonstrated close correlations between DFU and patients with calluses/corns (χ2 = 4.584, p = 0.0323), tinea pedis (χ2 = 4.030, p = 0.0447), and cracked skin (χ2 = 8.712, p = 0.0032). Only a small number of patients seek for the assistance from specialists, such as trimming toenails (3.4%), removing corn or calluses (1.4%) or treating wounds (11.78%), when they are suffering from foot problems. The findings of this study can potentially be utilized to develop an early DFU diagnostic method in diabetic patients and can provide objective evidence for suggesting that patients who are suffering from foot problems should seek professional help.


Asunto(s)
Amputación Quirúrgica/tendencias , Diabetes Mellitus Tipo 2/complicaciones , Pie Diabético/epidemiología , Diagnóstico Precoz , Exposición a Riesgos Ambientales/efectos adversos , Hospitales Urbanos/estadística & datos numéricos , Medición de Riesgo , Anciano , China/epidemiología , Diabetes Mellitus Tipo 2/diagnóstico , Pie Diabético/diagnóstico , Pie Diabético/etiología , Exposición a Riesgos Ambientales/estadística & datos numéricos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Estudios Retrospectivos , Factores de Riesgo
15.
J Invest Dermatol ; 137(3): 650-659, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27840235

RESUMEN

Disruption of the barrier function of skin increases transepidermal water loss and up-regulates inflammatory pathways in the epidermis. Consequently, sustained expression of proinflammatory cytokines from the epidermis is associated with dermal scarring. We found increased expression of S100A12 in the epidermis of human hypertrophic and keloid scar. Exposing a stratified keratinocyte culture to a reduced-hydration environment increased the expression and secretion of S100A12 by nearly 70%, which in turn activated dermal fibroblasts in vitro. Direct treatment of fibroblasts with conditioned medium collected from stratified keratinocyte culture under reduced-hydration conditions activated fibroblasts, shown by up-regulation of α-smooth muscle actin, pro-collagen 1, and F-actin expression. However, this fibroblast activation was not found when S100A12 was knocked down by RNA interference in keratinocytes. Pharmacological blockade of S100A12 receptors, RAGE, or TLR4 inhibited S100A12-induced fibroblast activation. Local delivery of S100A12 resulted in a marked hypertrophic scar formation in a validated rabbit hypertrophic scar model compared with saline control. Our findings indicate that S100A12 functions as a proinflammatory cytokine and suggest that S100A12 is a potential therapeutic target for dermal scarring.


Asunto(s)
Epidermis/metabolismo , Epidermis/patología , Fibroblastos/metabolismo , Proteína S100A12/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Cicatriz Hipertrófica/metabolismo , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Femenino , Fibrosis , Humanos , Inflamación , Queloide/metabolismo , Queratinocitos/citología , Interferencia de ARN , Conejos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor Toll-Like 4/metabolismo
16.
Plast Reconstr Surg ; 137(2): 534-543, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26818288

RESUMEN

BACKGROUND: Localized oxygen deficiency plays a central role in the pathogenesis of chronic wounds; thus, rectifying localized ischemia with oxygen therapy has been postulated to be an integral aspect of the management of chronic wounds. The efficacy of a novel approach for oxygen therapy on chronic wound healing was evaluated. METHODS: Oxygen was delivered to ischemic wounds by means of the topical application of oxygenated, chemically modified bovine hemoglobin (IKOR 2084) in a validated rabbit ear ischemic wound model. The wound healing was evaluated histologically by measuring epithelial gap and neo-granulation tissue area. In situ expression of endothelial cells (CD31) and proliferative cells (Ki-67) was examined by immunohistochemistry analysis. The mRNA of vascular endothelial growth factor, endothelial nitric oxide synthase, and matrix metalloproteinase-9 was quantified by real-time reverse-transcriptase polymerase chain reaction. The collagen was detected by Sirius red staining. RESULTS: In comparison with topical application of saline, the administration of oxygenated IKOR 2084 increases wound reepithelialization and formation of neo-granulation tissue in a dose-dependent manner, and cellular proliferation (Ki-67). Conversely, the administration of deoxygenated IKOR 2084 aggravated the ischemic wound healing process. Moreover, the topical administration of oxygenated IKOR 2084 induces angiogenesis as evidenced by concomitant increases in CD31 protein and vascular endothelial growth factor and endothelial nitric oxide synthase mRNA expression in treated wounds. Oxygenated IKOR 2084 administration also increased collagen deposition in wounds, with decreases in the expression of matrix metalloproteinase-9 mRNA. CONCLUSION: This study suggests that the topical application of oxygenated IKOR 2084 ameliorates the reparative progress of ischemic wounds through enhanced angiogenesis, cellular proliferation, and collagen deposition.


Asunto(s)
Oído/irrigación sanguínea , Hemoglobinas/administración & dosificación , Daño por Reperfusión/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Administración Tópica , Animales , Bovinos , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Conejos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Resultado del Tratamiento
17.
Am J Pathol ; 186(1): 109-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26597884

RESUMEN

The most critical function of the epidermis is to prevent water loss and maintain skin homeostasis. Disruption of the functional skin barrier causes delayed wound healing, hypertrophic scarring, and many skin diseases. Herein, we show that reduced hydration increases the expression of S100 protein family members, S100A8/S100A9, in stratified keratinocyte culture and human ex vivo skin culture. Immunohistological analyses show that S100A8/A9 are highly expressed in the epidermis of human hypertrophic scar and keloid tissues. Reduced hydration demonstrates activation of fibroblasts in the keratinocyte-fibroblast co-culture. In contrast, knockdown of S100A8 or S100A9 by RNA interference in keratinocytes failed to activate fibroblasts. Pretreatment with pharmacological blockers of S100A8/A9 receptors, Toll-like receptor 4 and receptor for advanced glycation end products, inhibits fibroblast activation induced by recombinant S100A8/A9 proteins. Moreover, we observe that local delivery of S100A8 protein results in a marked increase in hypertrophic scarring in the in vivo rabbit ear scar model. Our results indicate that hydration status promotes fibroblast activation and fibrosis by directly affecting the expression of inflammatory signaling in keratinocytes, thereby strongly suggesting S100A8/A9 to be novel targets in preventing scarring.


Asunto(s)
Calgranulina A/biosíntesis , Calgranulina B/biosíntesis , Cicatriz/metabolismo , Epidermis/patología , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Adulto , Animales , Western Blotting , Técnicas de Cocultivo , Dermis/patología , Femenino , Fibrosis/patología , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Agua , Adulto Joven
18.
Sci Transl Med ; 7(312): 312ra177, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26537257

RESUMEN

The mechanisms by which the epidermis responds to disturbances in barrier function and restores homeostasis are unknown. With a perturbation of the epidermal barrier, water is lost, resulting in an increase in extracellular sodium concentration. We demonstrate that the sodium channel Nax functions as a sodium sensor. With increased extracellular sodium, Nax up-regulates prostasin, which results in activation of the sodium channel ENaC, resulting in increased sodium flux and increased downstream mRNA synthesis of inflammatory mediators. Nax is present in multiple epithelial tissues, and up-regulation of its downstream genes is found in hypertrophic scars. In animal models, blocking Nax expression results in improvement in scarring and atopic dermatitis-like symptoms, both of which are pathological conditions characterized by perturbations in barrier function. These findings support an important role for Nax in maintaining epithelial homeostasis.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Queratinocitos/metabolismo , Serina Endopeptidasas/metabolismo , Sodio/metabolismo , Animales , Cicatriz/metabolismo , Cicatriz/patología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Homeostasis , Humanos , Activación del Canal Iónico , Queratinocitos/patología , Ratones Pelados , Conejos , Regulación hacia Arriba , Xenopus laevis
19.
J Invest Dermatol ; 135(3): 796-806, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25371970

RESUMEN

Although it is known that the inflammatory response that results from disruption of epithelial barrier function after injury results in excessive scarring, the upstream signals remain unknown. It has also been observed that epithelial disruption results in reduced hydration status and that the use of occlusive dressings that prevent water loss from wounds decreases scar formation. We hypothesized that hydration status changes sodium homeostasis and induces sodium flux in keratinocytes, which result in activation of pathways responsible for keratinocyte-fibroblast signaling and ultimately lead to activation of fibroblasts. Here, we demonstrate that perturbations in epithelial barrier function lead to increased sodium flux in keratinocytes. We identified that sodium flux in keratinocytes is mediated by epithelial sodium channels (ENaCs) and causes increased secretion of proinflammatory cytokines, which activate fibroblast via the cyclooxygenase 2 (COX-2)/prostaglandin E2 (PGE2) pathway. Similar changes in signal transduction and sodium flux occur by increased sodium concentration, which simulates reduced hydration, in the media in epithelial cultures or human ex vivo skin cultures. Blockade of ENaC, prostaglandin synthesis, or PGE2 receptors all reduce markers of fibroblast activation and collagen synthesis. In addition, employing a validated in vivo excessive scar model in the rabbit ear, we demonstrate that utilization of either an ENaC blocker or a COX-2 inhibitor results in a marked reduction in scarring. Other experiments demonstrate that the activation of COX-2 in response to increased sodium flux is mediated through the PIK3/Akt pathway. Our results indicate that ENaC responds to small changes in sodium concentration with inflammatory mediators and suggest that the ENaC pathway is a potential target for a strategy to prevent fibrosis.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Homeostasis/fisiología , Inflamación/metabolismo , Transducción de Señal/fisiología , Piel/metabolismo , Sodio/metabolismo , Agua/metabolismo , Animales , Comunicación Celular/fisiología , Células Cultivadas , Cicatriz/prevención & control , Técnicas de Cocultivo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/antagonistas & inhibidores , Dinoprostona/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/patología , Modelos Animales , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Conejos , Piel/efectos de los fármacos , Piel/patología
20.
Wound Repair Regen ; 22(6): 712-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25230854

RESUMEN

The importance of bacterial biofilms to chronic wound pathogenesis is well established. Different treatment modalities, including topical dressings, have yet to show consistent efficacy against wound biofilm. This study evaluates the impact of a novel, antimicrobial Test Dressing on Pseudomonas aeruginosa biofilm-infected wounds. Six-mm dermal punch wounds in rabbit ears were inoculated with 10(6) colony-forming units of P. aeruginosa. Biofilm was established in vivo using our published model. Dressing changes were performed every other day with either Active Control or Test Dressings. Treated and untreated wounds were harvested for several quantitative endpoints. Confirmatory studies were performed to measure treatment impact on in vitro P. aeruginosa and in vivo polybacterial wounds containing P. aeruginosa and Staphylococcus aureus. The Test Dressing consistently decreased P. aeruginosa bacterial counts, and improved wound healing relative to Inactive Vehicle and Active Control wounds (p < 0.05). In vitro bacterial counts were also significantly reduced following Test Dressing therapy (p < 0.05). Similarly, improvements in bacterial burden and wound healing were also achieved in polybacterial wounds (p < 0.05). This study represents the first quantifiable and consistent in vivo evidence of a topical antimicrobial dressing's impact against established wound biofilm. The development of clinically applicable therapies against biofilm such as this is critical to improving chronic wound care.


Asunto(s)
Antiinfecciosos Locales/farmacología , Vendajes , Biopelículas/efectos de los fármacos , Infecciones por Pseudomonas/terapia , Pseudomonas aeruginosa/efectos de los fármacos , Traumatismos de los Tejidos Blandos/microbiología , Infección de Heridas/microbiología , Infección de Heridas/terapia , Animales , Biopelículas/crecimiento & desarrollo , Modelos Animales de Enfermedad , Oído , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/crecimiento & desarrollo , Conejos , Traumatismos de los Tejidos Blandos/terapia , Cicatrización de Heridas , Infección de Heridas/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...